49 results on '"Clewell RA"'
Search Results
2. Comparison of Rat Hepatocyte 2D-Monocultures and Hepatocytes Non-Parenchymal Cell Co-Cultures for Assessing Chemical Toxicity.
- Author
-
Stefanowicz AJ, Recio L, Black MB, Beames T, Andersen ME, Stern RA, Clewell RA, McMullen PD, Hartman JK, and Ranade A
- Subjects
- Rats, Animals, Coculture Techniques, Acetaminophen toxicity, Models, Biological, Cells, Cultured, Hepatocytes metabolism, Liver metabolism
- Abstract
Liver responses are the most common endpoints used as the basis for setting exposure standards. Liver hepatocytes play a vital role in biotransformation of xenobiotics, but non-parenchymal cells (NPCs) in the liver are also involved in certain liver responses. Development of in vitro systems that more faithfully capture liver responses to reduce reliance on animals is a major focus of New Approach Methodology (NAMs). Since rodent regulatory studies are frequently the sole source safety assessment data, mode-of-action data, and used for risk assessments, in vitro rodent models that reflect in vivo responses need to be developed to reduce reliance on animal models. In the work presented in this paper, we developed a 2-D hepatocyte monoculture and 2-D liver cell co-culture system using rat liver cells. These models were assessed for conditions for short-term stability of the cultures and phenotypic and transcriptomic responses of 2 prototypic hepatotoxicants compounds - acetaminophen and phenobarbital. The optimized multi-cellular 2-D culture required use of freshly prepared hepatocytes and NPCs from a single rat, a 3:1 ratio of hepatocytes to NPCs and growth medium using 50% Complete Williams E medium (WEM) and 50% Endothelial Cell Medium (ECM). The transcriptomic responses of the 2 model systems to PB were compared to previous studies from TG-Gates on the gene expression changes in intact rats and the co-culture model responses were more representative of the in vivo responses. Transcriptomic read-outs promise to move beyond conventional phenotypic evaluations with these in vitro NAMs and provide insights about modes of action.
- Published
- 2023
- Full Text
- View/download PDF
3. Estimating provisional margins of exposure for data-poor chemicals using high-throughput computational methods.
- Author
-
Nicolas CI, Linakis MW, Minto MS, Mansouri K, Clewell RA, Yoon M, Wambaugh JF, Patlewicz G, McMullen PD, Andersen ME, and Clewell Iii HJ
- Abstract
Current computational technologies hold promise for prioritizing the testing of the thousands of chemicals in commerce. Here, a case study is presented demonstrating comparative risk-prioritization approaches based on the ratio of surrogate hazard and exposure data, called margins of exposure (MoEs). Exposures were estimated using a U.S. EPA's ExpoCast predictive model (SEEM3) results and estimates of bioactivity were predicted using: 1) Oral equivalent doses (OEDs) derived from U.S. EPA's ToxCast high-throughput screening program, together with in vitro to in vivo extrapolation and 2) thresholds of toxicological concern (TTCs) determined using a structure-based decision-tree using the Toxtree open source software. To ground-truth these computational approaches, we compared the MoEs based on predicted noncancer TTC and OED values to those derived using the traditional method of deriving points of departure from no-observed adverse effect levels (NOAELs) from in vivo oral exposures in rodents. TTC-based MoEs were lower than NOAEL-based MoEs for 520 out of 522 (99.6%) compounds in this smaller overlapping dataset, but were relatively well correlated with the same ( r
2 = 0.59). TTC-based MoEs were also lower than OED-based MoEs for 590 (83.2%) of the 709 evaluated chemicals, indicating that TTCs may serve as a conservative surrogate in the absence of chemical-specific experimental data. The TTC-based MoE prioritization process was then applied to over 45,000 curated environmental chemical structures as a proof-of-concept for high-throughput prioritization using TTC-based MoEs. This study demonstrates the utility of exploiting existing computational methods at the pre-assessment phase of a tiered risk-based approach to quickly, and conservatively, prioritize thousands of untested chemicals for further study., Competing Interests: Author ML and HC are employed by Ramboll US Consulting, Inc. Authors MM, PM, and MA were employed by Scitovation, LLC. Author RC is employed by 21st Century Tox Consulting. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer NM declared a past co‐authorship with one of the authors GP to the handling Editor., (Copyright © 2022 Nicolas, Linakis, Minto, Mansouri, Clewell, Yoon, Wambaugh, Patlewicz, McMullen, Andersen and Clewell III.)- Published
- 2022
- Full Text
- View/download PDF
4. An in vitro approach to determine the human relevance of anti-spermatogenic effects of 4-methylmorpholine 4-oxide, monohydrate (NMMO) in rat reproductive toxicity studies.
- Author
-
Clewell RA, Clewell HJ 3rd, Linakis MW, Easley CA 4th, Langmo JN, Salley J, Gentry R, and Rücker T
- Subjects
- Animals, Humans, Male, Morpholines, Rats, Seminiferous Tubules, Spermatids, Spermatocytes, Testis, Oxides, Spermatogenesis
- Abstract
Reduced sperm counts have been observed in male rats in an extended one generation reproductive toxicity study (EOGRTS, OECD 443) following repeated administration of 300 mg/kg/day N-Methylmorpholine N-oxide (NMMO). However, no adverse effects on reproductive organs have been reported in studies conducted with NMMO, and the mode of action (MOA) for the effects of NMMO on spermatogenesis is unknown, which complicates the interpretation of these data for human risk assessment. Here, a New Approach Method (NAM) strategy was used to evaluate NMMO MOA and compare interspecies susceptibility for anti-spermatogenic effects using organotypic in vitro assays combined with in vitro metabolism and in vitro to in vivo extrapolation (IVIVE) biokinetic modeling to compare predicted oral equivalent doses (OEDs) in human and rat. Dose-response data were collected in isolated germ cells and in an ex vivo seminiferous tubule model that recapitulates the interaction between the somatic environment and differentiating germ cells to account for potential direct and indirect effects on germ cells. With regard to direct spermatogenic effects, the human isolated germ cell model showed no toxicity at doses ≤300 μM (OED ≤ 86 mg/kg/day). With regard to indirect effects, the rat ex vivo model demonstrated dose-dependent decreases in secondary spermatocyte populations at OEDs ≥89 mg/kg/day, and reduced expression of RNAs specific to several stages of spermatogenesis (spermatogonia, pachytene spermatocytes, round spermatids) at OED = 267 mg/kg/day, consistent with in vivo observations. In contrast, the monkey ex vivo model did not show dose-dependent decreases in these same RNAs, and often demonstrated increased trends instead. These studies demonstrate clear quantitative and qualitative differences in the rat and primate response to NMMO. Furthermore, effects observed in the rat in vitro culture were not observed in the monkey at concentrations equivalent to in vivo doses of up to 1376 mg/kg/day, which is higher than the in vivo dose limit in the EOGRT study, indicating that the isolated findings on spermatogenesis in the rat studies are not likely to be relevant to humans., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
5. Considerations for Improving Metabolism Predictions for In Vitro to In Vivo Extrapolation.
- Author
-
Moreau M, Mallick P, Smeltz M, Haider S, Nicolas CI, Pendse SN, Leonard JA, Linakis MW, McMullen PD, Clewell RA, Clewell HJ, and Yoon M
- Abstract
High-throughput (HT) in vitro to in vivo extrapolation (IVIVE) is an integral component in new approach method (NAM)-based risk assessment paradigms, for rapidly translating in vitro toxicity assay results into the context of in vivo exposure. When coupled with rapid exposure predictions, HT-IVIVE supports the use of HT in vitro assays for risk-based chemical prioritization. However, the reliability of prioritization based on HT bioactivity data and HT-IVIVE can be limited as the domain of applicability of current HT-IVIVE is generally restricted to intrinsic clearance measured primarily in pharmaceutical compounds. Further, current approaches only consider parent chemical toxicity. These limitations occur because current state-of-the-art HT prediction tools for clearance and metabolite kinetics do not provide reliable data to support HT-IVIVE. This paper discusses current challenges in implementation of IVIVE for prioritization and risk assessment and recommends a path forward for addressing the most pressing needs and expanding the utility of IVIVE., Competing Interests: Authors MM, PM, MS, SH, CN, SP, PM and MY were employed by the company ScitoVation, LLC, author RC was employed by the company. 21st Century Tox Consulting and authors ML and HC were employed by the company Ramboll US Corp. The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Moreau, Mallick, Smeltz, Haider, Nicolas, Pendse, Leonard, Linakis, McMullen, Clewell, Clewell and Yoon.)
- Published
- 2022
- Full Text
- View/download PDF
6. Time-dependent genomic response in primary human uroepithelial cells exposed to arsenite for up to 60 days.
- Author
-
Efremenko A, Balbuena P, Clewell RA, Black M, Pluta L, Andersen ME, Gentry PR, Yager JW, and Clewell HJ
- Subjects
- Adult, Arsenites administration & dosage, Cell Proliferation drug effects, Cells, Cultured, Dose-Response Relationship, Drug, Epithelial Cells cytology, Female, Gene Expression Regulation drug effects, Genomics, Humans, Male, Middle Aged, NF-kappa B metabolism, Oligonucleotide Array Sequence Analysis, Oxidative Stress drug effects, Signal Transduction drug effects, Time Factors, Transcription Factor AP-1 metabolism, Up-Regulation drug effects, Ureter cytology, Ureter drug effects, Urothelium cytology, Young Adult, Arsenites toxicity, Epithelial Cells drug effects, Proto-Oncogene Proteins c-mdm2 genetics, Urothelium drug effects
- Abstract
Evidence from both in vivo and in vitro studies suggests that gene expression changes from long-term exposure to arsenite evolve markedly over time, including reversals in the direction of expression change in key regulatory genes. In this study, human uroepithelial cells from the ureter segments of 4 kidney-donors were continuously treated in culture with arsenite at concentrations of 0.1 or 1 μM for 60 days. Gene expression at 10, 20, 30, 40, and 60 days was determined using Affymetrix human genome microarrays and signal pathway analysis was performed using GeneGo Metacore. Arsenic treated cells continued to proliferate for the full 60-day period, whereas untreated cells ceased proliferating after approximately 30 days. A peak in the number of gene changes in the treated cells compared to untreated controls was observed between 30 and 40 days of exposure, with substantially fewer changes at 10 and 60 days, suggesting remodeling of the cells over time. Consistent with this possibility, the direction of expression change for a number of key genes was reversed between 20 and 30 days, including CFOS and MDM2. While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKα). At around 30 days, there was a transition to increased inflammatory and proliferative signaling (AKT, CFOS), evidence of epithelial-to-mesenchymal transition (EMT), and alterations in DNA damage responses (MDM2, ATM). A common element in the changing response of cells to arsenite over time appears to involve up-regulation of MDM2 by inflammatory signaling (through AP-1 and NF-κB), leading to inhibition of P53 function., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
7. Development of a physiologically based pharmacokinetic model of diisononyl phthalate (DiNP) in pregnant rat and human.
- Author
-
Campbell JL Jr, Otter R, Anderson WA, Longnecker MP, Clewell RA, North C, and Clewell HJ 3rd
- Subjects
- Animals, Female, Humans, Intestines, Liver metabolism, Metabolic Detoxication, Phase II, Models, Animal, Oxidation-Reduction, Plasma metabolism, Pregnancy, Rats, Environmental Pollutants pharmacokinetics, Phthalic Acids pharmacokinetics, Plasticizers pharmacokinetics
- Abstract
A physiologically based pharmacokinetic (PBPK) model for di-isononyl phthalate (DiNP) was developed by adapting the existing models for di(2-ethylhexyl) phthalate (DEHP) and di-butylphthalate (DBP). Both pregnant rat and human time-course plasma and urine data were used to address the hydrolysis of DiNP in intestinal tract, plasma, and liver as well as hepatic oxidative metabolism and conjugation of the monoester and primary oxidative metabolites. Data in both rats and humans were available to inform the uptake and disposition of mono-isononyl phthalate (MiNP) as well as the three primary oxidative metabolites including hydroxy (7-OH)-, oxo (7-OXO)-, and carboxy (7-COX)-monoisononyl phthalate in plasma and urine. The DiNP model was reliable over a wide range of exposure levels in the pregnant rat as well as the two low exposure levels in humans including capturing the nonlinear behavior in the pregnant rat after repeated 750 mg/kg/day dosing. The presented DiNP PBPK model in pregnant rat and human, based upon an extensive kinetic dataset in both species, may provide a basis for assessing human equivalent exposures based upon either rodent or in vitro points of departure.
- Published
- 2020
- Full Text
- View/download PDF
8. Application of a combined aggregate exposure pathway and adverse outcome pathway (AEP-AOP) approach to inform a cumulative risk assessment: A case study with phthalates.
- Author
-
Clewell RA, Leonard JA, Nicolas CI, Campbell JL, Yoon M, Efremenko AY, McMullen PD, Andersen ME, Clewell HJ 3rd, Phillips KA, and Tan YM
- Subjects
- Adverse Outcome Pathways, Animals, Humans, Male, Rats, Sexual Development drug effects, Dibutyl Phthalate pharmacokinetics, Dibutyl Phthalate pharmacology, Dibutyl Phthalate toxicity, Diethylhexyl Phthalate pharmacokinetics, Diethylhexyl Phthalate pharmacology, Diethylhexyl Phthalate toxicity, Environmental Exposure adverse effects, Environmental Pollutants pharmacokinetics, Environmental Pollutants pharmacology, Environmental Pollutants toxicity, Models, Biological
- Abstract
Advancements in measurement and modeling capabilities are providing unprecedented access to estimates of chemical exposure and bioactivity. With this influx of new data, there is a need for frameworks that help organize and disseminate information on chemical hazard and exposure in a manner that is accessible and transparent. A case study approach was used to demonstrate integration of the Adverse Outcome Pathway (AOP) and Aggregate Exposure Pathway (AEP) frameworks to support cumulative risk assessment of co-exposure to two phthalate esters that are ubiquitous in the environment and that are associated with disruption of male sexual development in the rat: di(2-ethylhexyl) phthalate (DEHP) and di-n-butyl phthalate (DnBP). A putative AOP was developed to guide selection of an in vitro assay for derivation of bioactivity values for DEHP and DnBP and their metabolites. AEPs for DEHP and DnBP were used to extract key exposure data as inputs for a physiologically based pharmacokinetic (PBPK) model to predict internal metabolite concentrations. These metabolite concentrations were then combined using in vitro-based relative potency factors for comparison with an internal dose metric, resulting in an estimated margin of safety of ~13,000. This case study provides an adaptable workflow for integrating exposure and toxicity data by coupling AEP and AOP frameworks and using in vitro and in silico methodologies for cumulative risk assessment., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
9. Identifying qualitative differences in PPARα signaling networks in human and rat hepatocytes and their significance for next generation chemical risk assessment methods.
- Author
-
McMullen PD, Bhattacharya S, Woods CG, Pendse SN, McBride MT, Soldatow VY, Deisenroth C, LeCluyse EL, Clewell RA, and Andersen ME
- Subjects
- Animals, Butyrates pharmacology, Cells, Cultured, Gene Expression Regulation drug effects, Hepatocytes drug effects, Humans, Male, PPAR alpha agonists, PPAR alpha genetics, Phenylurea Compounds pharmacology, Rats, Sprague-Dawley, Signal Transduction drug effects, Hepatocytes metabolism, PPAR alpha metabolism, Risk Assessment methods
- Abstract
In this paper, we evaluate the PPARα signaling network in rats, examining transcriptional responses in primary hepatocytes exposed to a PPARα specific ligand, GW7647. These transcriptomic studies were complemented with ChIP-seq studies of PPARα binding and transcription binding motif identification for PPARα responsive genes. We also conducted a limited study of GW7647 dosing the in intact rat to examine differences in transcriptional responses for primary hepatocytes in vitro and in the intact liver. The rat network has a much larger number of down-regulated genes and pathways than we had found in the human and the PPARα binding motifs in rat differed for upregulated and down regulated genes. Based on these results and comparison with our previous work with the human PPARα signaling network, we identified qualitative differences in the transcriptional networks controlled by PPARα activation in the two species that provide an explanation of the interspecies differences in the responses of humans and rodents to GW7647 and likely to other PPARα agonists. These studies also allow some observations on the manner in which in vitro, fit-for-purpose assays in human hepatocytes could form the basis for risk assessment without recourse to in-life studies in rodents or other test species., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
10. The role of fit-for-purpose assays within tiered testing approaches: A case study evaluating prioritized estrogen-active compounds in an in vitro human uterotrophic assay.
- Author
-
Beames T, Moreau M, Roberts LA, Mansouri K, Haider S, Smeltz M, Nicolas CI, Doheny D, Phillips MB, Yoon M, Becker RA, McMullen PD, Andersen ME, Clewell RA, and Hartman JK
- Subjects
- Animals, Biological Assay methods, Cell Culture Techniques, Cell Line, Tumor, Cell Proliferation drug effects, Computer Simulation, Feasibility Studies, Female, Humans, Models, Biological, Rats, Risk Assessment methods, Uterus cytology, Animal Use Alternatives methods, Endocrine Disruptors toxicity, High-Throughput Screening Assays methods, Toxicity Tests methods, Uterus drug effects
- Abstract
Chemical risk assessment relies on toxicity tests that require significant numbers of animals, time and costs. For the >30,000 chemicals in commerce, the current scale of animal testing is insufficient to address chemical safety concerns as regulatory and product stewardship considerations evolve to require more comprehensive understanding of potential biological effects, conditions of use, and associated exposures. We demonstrate the use of a multi-level new approach methodology (NAMs) strategy for hazard- and risk-based prioritization to reduce animal testing. A Level 1/2 chemical prioritization based on estrogen receptor (ER) activity and metabolic activation using ToxCast data was used to select 112 chemicals for testing in a Level 3 human uterine cell estrogen response assay (IKA assay). The Level 3 data were coupled with quantitative in vitro to in vivo extrapolation (Q-IVIVE) to support bioactivity determination (as a surrogate for hazard) in a tissue-specific context. Assay AC
50 s and Q-IVIVE were used to estimate human equivalent doses (HEDs), and HEDs were compared to rodent uterotrophic assay in vivo-derived points of departure (PODs). For substances active both in vitro and in vivo, IKA assay-derived HEDs were lower or equivalent to in vivo PODs for 19/23 compounds (83%). Activity exposure relationships were calculated, and the IKA assay was as or more protective of human health than the rodent uterotrophic assay for all IKA-positive compounds. This study demonstrates the utility of biologically relevant fit-for-purpose assays and supports the use of a multi-level strategy for chemical risk assessment., Competing Interests: Declaration of competing interest None., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
11. Addressing systematic inconsistencies between in vitro and in vivo transcriptomic mode of action signatures.
- Author
-
McMullen PD, Pendse SN, Black MB, Mansouri K, Haider S, Andersen ME, and Clewell RA
- Subjects
- Animals, Databases, Factual, Gene Ontology, Hepatocytes metabolism, Humans, Risk Assessment, Transcriptome, Gene Expression Profiling
- Abstract
Because of their broad biological coverage and increasing affordability transcriptomic technologies have increased our ability to evaluate cellular response to chemical stressors, providing a potential means of evaluating chemical response while decreasing dependence on apical endpoints derived from traditional long-term animal studies. It has recently been suggested that dose-response modeling of transcriptomic data may be incorporated into risk assessment frameworks as a means of approximating chemical hazard. However, identification of mode of action from transcriptomics lacks a similar systematic framework. To this end, we developed a web-based interactive browser-MoAviz-that allows visualization of perturbed pathways. We populated this browser with expression data from a large public toxicogenomic database (TG-GATEs). We evaluated the extent to which gene expression changes from in-life exposures could be associated with mode of action by developing a novel similarity index-the Modified Jaccard Index (MJI)-that provides a quantitative description of genomic pathway similarity (rather than gene level comparison). While typical compound-compound similarity is low (median MJI = 0.026), clustering of the TG-GATES compounds identifies groups of similar chemistries. Some clusters aggregated compounds with known similar modes of action, including PPARa agonists (median MJI = 0.315) and NSAIDs (median MJI = 0.322). Analysis of paired in vitro (hepatocyte)-in vivo (liver) experiments revealed systematic patterns in the responses of model systems to chemical stress. Accounting for these model-specific, but chemical-independent, differences improved pathway concordance by 36% between in vivo and in vitro models., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
12. An evaluation of the USEPA Proposed Approaches for applying a biologically based dose-response model in a risk assessment for perchlorate in drinking water.
- Author
-
Clewell HJ 3rd, Gentry PR, Hack CE, Greene T, and Clewell RA
- Subjects
- Dose-Response Relationship, Drug, Humans, Risk Assessment, United States, United States Environmental Protection Agency, Drinking Water chemistry, Perchlorates analysis, Water Pollutants, Chemical analysis
- Abstract
The United States Environmental Protection Agency's (USEPA) 2017 report, "Draft Report: Proposed Approaches to Inform the Derivation of a Maximum Contaminant Level Goal for Perchlorate in Drinking Water", proposes novel approaches for deriving a Maximum Contaminant Level Goal (MCLG) for perchlorate using a biologically-based dose-response (BBDR) model. The USEPA (2017) BBDR model extends previously peer-reviewed perchlorate models to describe the relationship between perchlorate exposure and thyroid hormone levels during early pregnancy. Our evaluation focuses on two key elements of the USEPA (2017) report: the plausibility of BBDR model revisions to describe control of thyroid hormone production in early pregnancy and the basis for linking BBDR model results to neurodevelopmental outcomes. While the USEPA (2017) BBDR model represents a valuable research tool, the lack of supporting data for many of the model assumptions and parameters calls into question the fitness of the extended BBDR model to support quantitative analyses for regulatory decisions on perchlorate in drinking water. Until more data can be developed to address uncertainties in the current BBDR model, USEPA should continue to rely on the RfD recommended by the NAS (USEPA, 2005) when considering further regulatory action., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. Dose-dependence of chemical carcinogenicity: Biological mechanisms for thresholds and implications for risk assessment.
- Author
-
Clewell RA, Thompson CM, and Clewell HJ 3rd
- Subjects
- Animals, Dose-Response Relationship, Drug, Humans, Risk Assessment, Social Control, Formal, Carcinogens toxicity
- Abstract
Current regulatory practices for chemical carcinogens were established when scientific understanding of the molecular mechanisms of chemical carcinogenesis was in its infancy. Initial discovery that DNA mutation was the root of cancer led quickly to regulatory processes that assumed such a simple relationship could be described with a linear approach. This linear, no threshold approach has since become the default approach to risk assessment of chemicals with carcinogenic potential. Since then, a multitude of intrinsic processes have been identified at the molecular, cellular and organism level that work to prevent transient DNA damage from causing permanent mutations, and mutated cells from becoming cancer. Mounting evidence indicates that these protective mechanisms can prevent carcinogenesis at low doses of genotoxic chemicals, leading to non-linear dose-response. Further, a number of non-genotoxic mechanisms have demonstrated threshold-shaped dose-response for cancer outcomes. The existence of non-linear dose-response curves for both non-genotoxic and genotoxic chemical carcinogens stands in stark contrast to the default risk assessment approach that assumes low dose linearity. In this review, we highlight some of the key discoveries and technological advances that have influenced scientific understanding of chemical carcinogenesis over the last fifty years and provide case studies to demonstrate the utility of these modern technologies in providing a biologically robust evaluation of chemical dose-response for cancer risk assessment., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
14. Assessing bioactivity-exposure profiles of fruit and vegetable extracts in the BioMAP profiling system.
- Author
-
Wetmore BA, Clewell RA, Cholewa B, Parks B, Pendse SN, Black MB, Mansouri K, Haider S, Berg EL, Judson RS, Houck KA, Martin M, Clewell HJ 3rd, Andersen ME, Thomas RS, and McMullen PD
- Subjects
- Biological Assay, Cells, Cultured, Food, Organic, Humans, Metals, Heavy analysis, Metals, Heavy toxicity, Mycotoxins analysis, Mycotoxins toxicity, Pesticide Residues analysis, Pesticide Residues toxicity, Plant Extracts analysis, Toxicity Tests, Fruit, High-Throughput Screening Assays, Magnoliopsida, Plant Extracts toxicity, Vegetables
- Abstract
The ToxCast program has generated in vitro screening data on over a thousand chemicals to assess potential disruption of important biological processes and assist in hazard identification and chemical testing prioritization. Few results have been reported for complex mixtures. To extend these ToxCast efforts to mixtures, we tested extracts from 30 organically grown fruits and vegetables in concentration-response in the BioMAP® assays. BioMAP systems use human primary cells primed with endogenous pathway activators to identify phenotypic perturbations related to proliferation, inflammation, immunomodulation, and tissue remodeling. Clustering of bioactivity profiles revealed separation of these produce extracts and ToxCast chemicals. Produce extracts elicited 87 assay endpoint responses per item compared to 20 per item for ToxCast chemicals. On a molar basis, the produce extracts were 10 to 50-fold less potent and when constrained to the maximum testing concentration of the ToxCast chemicals, the produce extracts did not show activity in as many assay endpoints. Using intake adjusted measures of dose, the bioactivity potential was higher for produce extracts than for agrichemicals, as expected based on the comparatively small amounts of agrichemical residues present on conventionally grown produce. The evaluation of BioMAP readouts and the dose responses for produce extracts showed qualitative and quantitative differences from results with single chemicals, highlighting challenges in the interpretation of bioactivity data and dose-response from complex mixtures., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
15. Developing context appropriate toxicity testing approaches using new alternative methods (NAMs).
- Author
-
Andersen ME, McMullen PD, Phillips MB, Yoon M, Pendse SN, Clewell HJ, Hartman JK, Moreau M, Becker RA, and Clewell RA
- Subjects
- Animals, Computational Biology methods, Computational Chemistry methods, High-Throughput Screening Assays, Humans, In Vitro Techniques, Mammals, Animal Testing Alternatives trends, Toxicity Tests trends
- Abstract
In the past 10 years, the public, private, and non-profit sectors have found agreement that hazard identification and risk assessment should capitalize on the explosion of knowledge in the biological sciences, moving away from in life animal testing toward more human-relevant in vitro and in silico methods, collectively referred to as new approach methodologies (NAMs). The goals for implementation of NAMs are to efficiently identify possible chemical hazards and to gather dose-response data to inform more human-relevant safety assessment. While work proceeds to develop NAMs, there has been less emphasis on creating decision criteria or showing how risk context should guide selection and use of NAMs. Here, we outline application scenarios for NAMs in different risk contexts and place different NAMs and conventional testing approaches into four broad levels. Level 1 relies solely on computational screening; Level 2 consists of high throughput in vitro screening with human cells intended to provide broad coverage of possible responses; Level 3 focuses on fit-for-purpose assays selected based on presumptive modes of action (MOA) and designed to provide more quantitative estimates of relevant dose responses; Level 4 has a variety of more complex multi-dimensional or multi-cellular assays and might include targeted in vivo studies to further define MOA. Each level also includes decision-appropriate exposure assessment tools. Our aims here are to (1) foster discussion about context-dependent applications of NAMs in relation to risk assessment needs and (2) describe a functional roadmap to identify where NAMs are expected to be adequate for chemical safety decision-making.
- Published
- 2019
- Full Text
- View/download PDF
16. A Qualitative Modeling Approach for Whole Genome Prediction Using High-Throughput Toxicogenomics Data and Pathway-Based Validation.
- Author
-
Haider S, Black MB, Parks BB, Foley B, Wetmore BA, Andersen ME, Clewell RA, Mansouri K, and McMullen PD
- Abstract
Efficient high-throughput transcriptomics (HTT) tools promise inexpensive, rapid assessment of possible biological consequences of human and environmental exposures to tens of thousands of chemicals in commerce. HTT systems have used relatively small sets of gene expression measurements coupled with mathematical prediction methods to estimate genome-wide gene expression and are often trained and validated using pharmaceutical compounds. It is unclear whether these training sets are suitable for general toxicity testing applications and the more diverse chemical space represented by commercial chemicals and environmental contaminants. In this work, we built predictive computational models that inferred whole genome transcriptional profiles from a smaller sample of surrogate genes. The model was trained and validated using a large scale toxicogenomics database with gene expression data from exposure to heterogeneous chemicals from a wide range of classes (the Open TG-GATEs data base). The method of predictor selection was designed to allow high fidelity gene prediction from any pre-existing gene expression data set, regardless of animal species or data measurement platform. Predictive qualitative models were developed with this TG-GATES data that contained gene expression data of human primary hepatocytes with over 941 samples covering 158 compounds. A sequential forward search-based greedy algorithm, combining different fitting approaches and machine learning techniques, was used to find an optimal set of surrogate genes that predicted differential expression changes of the remaining genome. We then used pathway enrichment of up-regulated and down-regulated genes to assess the ability of a limited gene set to determine relevant patterns of tissue response. In addition, we compared prediction performance using the surrogate genes found from our greedy algorithm (referred to as the SV2000) with the landmark genes provided by existing technologies such as L1000 (Genometry) and S1500 (Tox21), finding better predictive performance for the SV2000. The ability of these predictive algorithms to predict pathway level responses is a positive step toward incorporating mode of action (MOA) analysis into the high throughput prioritization and testing of the large number of chemicals in need of safety evaluation.
- Published
- 2018
- Full Text
- View/download PDF
17. An in vitro approach for prioritization and evaluation of chemical effects on glucocorticoid receptor mediated adipogenesis.
- Author
-
Hartman JK, Beames T, Parks B, Doheny D, Song G, Efremenko A, Yoon M, Foley B, Deisenroth C, McMullen PD, and Clewell RA
- Subjects
- Adipocytes drug effects, Adipokines metabolism, Cell Differentiation drug effects, Cell Survival drug effects, Cells, Cultured, Dexamethasone pharmacology, Fatty Acid-Binding Proteins biosynthesis, Gene Expression drug effects, Humans, L-Lactate Dehydrogenase metabolism, Lipid Metabolism drug effects, Receptors, Glucocorticoid agonists, Receptors, Glucocorticoid antagonists & inhibitors, Stem Cells drug effects, Adipogenesis drug effects, Receptors, Glucocorticoid drug effects
- Abstract
Rising obesity rates worldwide have socio-economic ramifications. While genetics, diet, and lack of exercise are major contributors to obesity, environmental factors may enhance susceptibility through disruption of hormone homeostasis and metabolic processes. The obesogen hypothesis contends that chemical exposure early in development may enhance adipocyte differentiation, thereby increasing the number of adipocytes and predisposing for obesity and metabolic disease. We previously developed a primary human adipose stem cell (hASC) assay to evaluate the effect of environmental chemicals on PPARG-dependent adipogenesis. Here, the assay was modified to determine the effects of chemicals on the glucocorticoid receptor (GR) pathway. In differentiation cocktail lacking the glucocorticoid agonist dexamethasone (DEX), hASCs do not differentiate into adipocytes. In the presence of GR agonists, adipocyte maturation was observed using phenotypic makers for lipid accumulation, adipokine secretion, and expression of key genes. To evaluate the role of environmental compounds on adipocyte differentiation, progenitor cells were treated with 19 prioritized compounds previously identified by ToxPi as having GR-dependent bioactivity, and multiplexed assays were used to confirm a GR-dependent mode of action. Five chemicals were found to be strong agonists. The assay was also modified to evaluate GR-antagonists, and 8/10 of the hypothesized antagonists inhibited adipogenesis. The in vitro bioactivity data was put into context with extrapolated human steady state concentrations (Css) and clinical exposure data (Cmax). These data support using a human adipose-derived stem cell differentiation assay to test the potential of chemicals to alter human GR-dependent adipogenesis., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
18. Evaluating opportunities for advancing the use of alternative methods in risk assessment through the development of fit-for-purpose in vitro assays.
- Author
-
McMullen PD, Andersen ME, Cholewa B, Clewell HJ 3rd, Dunnick KM, Hartman JK, Mansouri K, Minto MS, Nicolas CI, Phillips MB, Slattery S, Yoon M, and Clewell RA
- Subjects
- Animal Testing Alternatives, Animals, Government Regulation, Humans, In Vitro Techniques, Lung Diseases chemically induced, Lung Diseases pathology, Risk Assessment methods, Toxicity Tests methods
- Abstract
An evolving regulatory, scientific, and legislative landscape is driving a fundamental change in how chemical safety decisions are made. As we move to implement changes, regulatory agencies and industry are beginning to adopt tiered approaches, which leverage high-throughput screening technologies for prioritization and read across, followed by interrogation of "hit chemicals" with more rigorous dose-response assessment either in fit-for-purpose human cell-based assays or with traditional in vivo tests. However, to date, suitable in vitro alternatives do not exist for the vast majority of the organ toxicities that form the basis of current regulatory decisions. To successfully support safety decisions, biologically relevant, quantitative, cell-based assays that evaluate dose-response and identify regions of safety for chemical exposure are required. This review evaluates the current state of the science in the development of such assays, identifies key gaps in the current tests, and recommends areas where research efforts may be focused to help move the risk assessment community towards more wide-spread use of in vitro methods. Our analysis suggests that a key shortcoming in the current efforts is the ability to test volatile compounds and to predict pulmonary toxicity. We present a mechanistically-based path forward for the development of a fit-for-purpose lung toxicity assay., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts.
- Author
-
Miller MM, McMullen PD, Andersen ME, and Clewell RA
- Subjects
- Androgens, Biological Assay, Humans, Receptors, Estrogen metabolism, Risk Assessment methods, Toxicity Tests, United States, Endocrine Disruptors toxicity, Estrogens toxicity
- Abstract
Current in life toxicity testing paradigms are being challenged as the future of risk assessment moves towards more comprehensive mode of action/adverse outcome pathway based approaches. In particular, endocrine disruption screening is now a global activity and key initiatives in the United States focus on the use of high throughput in vitro assays to prioritize compounds for further testing of estrogen, androgen or thyroid disruption. Of these pathways, much of the emphasis to date has been on high-throughput methods for estrogenic activity primarily using ligand binding and trans-activation assays. However, as the knowledge regarding estrogen receptor signaling pathways continues to evolve, it is clear that the assumption of a simple one-receptor pathway underlying current in vitro screening assays is out of date. To develop more accurate models for estrogen-initiated pathways useful for quantitative safety assessments, we must design assays that account for the key signaling processes driving cellular dose response based on up-to-date understanding of the biological network. In this review, we summarize the state of the science for the estrogen receptor signaling network, particularly with regard to proliferative effects, and highlight gaps in current high throughput approaches. From the sum of this literature, we propose a model for the estrogen-signaling pathway that should serve as a starting point for development of new in vitro methods fit for the purpose of predicting dose response for estrogenic chemicals in the human.
- Published
- 2017
- Full Text
- View/download PDF
20. Contribution of ATM and ATR kinase pathways to p53-mediated response in etoposide and methyl methanesulfonate induced DNA damage.
- Author
-
Sun B, Ross SM, Rowley S, Adeleye Y, and Clewell RA
- Subjects
- Cell Cycle drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Flow Cytometry, Gene Knockdown Techniques, HCT116 Cells, Humans, Micronucleus Tests, Signal Transduction, Tumor Suppressor Protein p53 genetics, Apoptosis drug effects, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Damage, Etoposide toxicity, Mesylates toxicity, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 is a key integrator of cellular response to DNA damage, supporting post-translational repair and driving transcription-mediated responses including cell cycle arrest, apoptosis, and repair. DNA damage sensing kinases recognize different types of DNA damage and initiate specific responses through various post-translational modifications of p53. This study evaluated chemical specificity of the p53 pathway response by manipulating p53 or its upstream kinases and assessing the effect on DNA damage and cellular responses to prototype chemicals: etoposide (ETP, topoisomerase II inhibitor) and methyl methane sulfonate (MMS, alkylating agent). p53-deficient cells demonstrated reduced accumulation of the p53 target proteins MDM2, p21, and Wip1; reduced apoptotic response; and increased DNA damage (p-H2AX and micronuclei) with both chemicals. However, p53 was not essential for cell cycle arrest in HT1080 or HCT116 cells. The two chemicals induced different patterns of kinase activation, particularly in terms of Chk 1, Chk 2, p38, and ERK 1/2. However, inhibition of the ATM pathway showed a greater effect on p53 activtation, apoptosis, and accumulation of DNA damage than ATR-Chk 1 or the MAP kinases regardless of the chemical used. These results indicate that ATM is the predominant upstream kinase responsible for activation of the p53-mediated DNA damage response for both MMS and ETP, though the downstream kinase response is markedly different. Environ. Mol. Mutagen. 58:72-83, 2017. © 2017 Wiley Periodicals, Inc., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
21. Editor's Highlight: Screening ToxCast Prioritized Chemicals for PPARG Function in a Human Adipose-Derived Stem Cell Model of Adipogenesis.
- Author
-
Foley B, Doheny DL, Black MB, Pendse SN, Wetmore BA, Clewell RA, Andersen ME, and Deisenroth C
- Subjects
- Adiponectin metabolism, Adipose Tissue cytology, Adult, Humans, Middle Aged, PPAR gamma genetics, RNA, Small Interfering genetics, Adipogenesis, Adipose Tissue drug effects, Models, Biological, PPAR gamma physiology, Stem Cells cytology
- Abstract
The developmental origins of obesity hypothesis posits a multifaceted contribution of factors to the fetal origins of obesity and metabolic disease. Adipocyte hyperplasia in gestation and early childhood may result in predisposition for obesity later in life. Rodent in vitro and in vivo studies indicate that some chemicals may directly affect adipose progenitor cell differentiation, but the human relevance of these findings is unclear. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARG) is the master regulator of adipogenesis. Human adipose-derived stem cells (hASC) isolated from adipose tissue express endogenous isoforms of PPARG and represent a biologically relevant cell-type for evaluating activity of PPARG ligands. Here, a multi-endpoint approach based on a phenotypic adipogenesis assay was applied to screen a set of 60 chemical compounds identified in ToxCast Phase I as PPARG active (49) or inactive (11). Chemicals showing activity in the adipogenesis screen were further evaluated in a series of 4 orthogonal assays representing 7 different key events in PPARG-dependent adipogenesis, including gene transcription, protein expression, and adipokine secretion. An siRNA screen was also used to evaluate PPARG-dependence of the adipogenesis phenotype. A universal concentration-response design enabled inter-assay comparability and implementation of a weight-of-evidence approach for bioactivity classification. Collectively, a total of 14/49 (29%) prioritized chemicals were identified with moderate-to-strong activity for human adipogenesis. These results provide the first integrated screening approach of prioritized ToxCast chemicals in a human stem cell model of adipogenesis and provide insight into the capacity of PPARG-activating chemicals to modulate early life programming of adipose tissue., (© The Author 2016. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2017
- Full Text
- View/download PDF
22. Editor's Highlight: Development of an In vitro Assay Measuring Uterine-Specific Estrogenic Responses for Use in Chemical Safety Assessment.
- Author
-
Miller MM, Alyea RA, LeSommer C, Doheny DL, Rowley SM, Childs KM, Balbuena P, Ross SM, Dong J, Sun B, Andersen MA, and Clewell RA
- Subjects
- Cell Line, Tumor, Estrogens toxicity, Ethinyl Estradiol metabolism, Female, Humans, Uterus cytology, Epithelial Cells drug effects, Receptors, Estrogen metabolism, Uterus drug effects, Xenobiotics toxicity
- Abstract
A toxicity pathway approach was taken to develop an in vitro assay using human uterine epithelial adenocarcinoma (Ishikawa) cells as a replacement for measuring an in vivo uterotrophic response to estrogens. The Ishikawa cell was determined to be fit for the purpose of recapitulating in vivo uterine response by verifying fidelity of the biological pathway components and the dose-response predictions to women of child-bearing age. Expression of the suite of estrogen receptors that control uterine proliferation (ERα66, ERα46, ERα36, ERβ, G-protein coupled estrogen receptor (GPER)) were confirmed across passages and treatment conditions. Phenotypic responses to ethinyl estradiol (EE) from transcriptional activation of ER-mediated genes, to ALP enzyme induction and cellular proliferation occurred at concentrations consistent with estrogenic activity in adult women (low picomolar). To confirm utility of this model to predict concentration-response for uterine proliferation with xenobiotics, we tested the concentration-response for compounds with known uterine estrogenic activity in humans and compared the results to assays from the ToxCast and Tox21 suite of estrogen assays. The Ishikawa proliferation assay was consistent with in vivo responses and was a more sensitive measure of uterine response. Because this assay was constructed by first mapping the key molecular events for cellular response, and then ensuring that the assay incorporated these events, the resulting cellular assay should be a reliable tool for identifying estrogenic compounds and may provide improved quantitation of chemical concentration response for in vitro-based safety assessments., (© The Author 2016. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2016
- Full Text
- View/download PDF
23. Approaches for characterizing threshold dose-response relationships for DNA-damage pathways involved in carcinogenicity in vivo and micronuclei formation in vitro.
- Author
-
Clewell RA and Andersen ME
- Subjects
- Animals, Cell Line, Computational Biology, DNA drug effects, DNA metabolism, Dose-Response Relationship, Drug, Etoposide toxicity, Formaldehyde toxicity, Gene Expression Regulation, Humans, Methyl Methanesulfonate toxicity, Micronucleus Tests, Models, Animal, Models, Biological, Signal Transduction drug effects, Carcinogenesis, DNA Damage, DNA Repair drug effects, Mutagens toxicity
- Abstract
Assessing the shape of dose-response curves for DNA-damage in cellular systems and for the consequences of DNA damage in intact animals remains a controversial topic. This overview looks at aspects of the pharmacokinetics (PK) and pharmacodynamics (PD) of cellular DNA-damage/repair and their role in defining the shape of dose-response curves using an in vivo example with formaldehyde and in vitro examples for micronuclei (MN) formation with several test compounds. Formaldehyde is both strongly mutagenic and an endogenous metabolite in cells. With increasing inhaled concentrations, there were transitions in gene changes, from activation of selective stress pathway genes at low concentrations, to activation of pathways for cell-cycle control, p53-DNA damage, and stem cell niche pathways at higher exposures. These gene expression changes were more consistent with dose-dependent transitions in the PD responses to formaldehyde in epithelial cells in the intact rat rather than the low-dose linear extrapolation methods currently used for carcinogens. However, more complete PD explanations of non-linear dose response for creation of fixed damage in cells require detailed examination of cellular responses in vitro using measures of DNA damage and repair that are not easily accessible in the intact animal. In the second section of the article, we illustrate an approach from our laboratory that develops fit-for-purpose, in vitro assays and evaluates the PD of DNA damage and repair through studies using prototypical DNA-damaging agents. Examination of a broad range of responses in these cells showed that transcriptional upregulation of cell cycle control and DNA repair pathways only occurred at doses higher than those causing overt damage fixed damage-measured as MN formation. Lower levels of damage appear to be handled by post-translational repair process using pre-existing proteins. In depth evaluation of the PD properties of one such post-translational process (formation of DNA repair centers; DRCs) has indicated that the formation of DRCs and their ability to complete repair before replication are consistent with threshold behaviours for mutagenesis and, by extension, with chemical carcinogenesis., (© The Author 2016. Published by Oxford University Press on behalf of the UK Environmental Mutagen Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
24. Pathway Based Toxicology and Fit-for-Purpose Assays.
- Author
-
Clewell RA, McMullen PD, Adeleye Y, Carmichael PL, and Andersen ME
- Subjects
- Animal Testing Alternatives, Animals, DNA Damage, High-Throughput Screening Assays, Humans, In Vitro Techniques, PPAR alpha physiology, Proto-Oncogene Proteins c-mdm2 physiology, Tumor Suppressor Protein p53 physiology, Toxicity Tests methods, Toxicology
- Abstract
The field of toxicity testing for non-pharmaceutical chemicals is in flux with multiple initiatives in North America and the EU to move away from animal testing to mode-of-action based in vitro assays. In this arena, there are still obstacles to overcome, such as developing appropriate cellular assays, creating pathway-based dose-response models and refining in vitro-in vivo extrapolation (IVIVE) tools. Overall, it is necessary to provide assurances that these new approaches are adequately protective of human and ecological health. Another major challenge for individual scientists and regulatory agencies is developing a cultural willingness to shed old biases developed around animal tests and become more comfortable with mode-of-action based assays in human cells. At present, most initiatives focus on developing in vitro alternatives and assessing how well these alternative methods reproduce past results related to predicting organism level toxicity in intact animals. The path forward requires looking beyond benchmarking against high dose animal studies. We need to develop targeted cellular assays, new cell biology-based extrapolation models for assessing regions of safety for chemical exposures in human populations, and mode-of-action-based approaches which are constructed on an understanding of human biology. Furthermore, it is essential that assay developers have the flexibility to 'validate' against the most appropriate mode-of-action data rather than against apical endpoints in high dose animal studies. This chapter demonstrates the principles of fit-for-purpose assay development using pathway-targeted case studies. The projects include p53-mdm2-mediated DNA-repair, estrogen receptor-mediated cell proliferation and PPARα receptor-mediated liver responses.
- Published
- 2016
- Full Text
- View/download PDF
25. Contributions of DNA repair and damage response pathways to the non-linear genotoxic responses of alkylating agents.
- Author
-
Klapacz J, Pottenger LH, Engelward BP, Heinen CD, Johnson GE, Clewell RA, Carmichael PL, Adeleye Y, and Andersen ME
- Subjects
- Alkylation genetics, Apoptosis genetics, DNA Adducts drug effects, Dose-Response Relationship, Drug, Humans, Mutagenicity Tests methods, Alkylating Agents toxicity, DNA Adducts genetics, DNA Damage genetics, DNA Repair genetics
- Abstract
From a risk assessment perspective, DNA-reactive agents are conventionally assumed to have genotoxic risks at all exposure levels, thus applying a linear extrapolation for low-dose responses. New approaches discussed here, including more diverse and sensitive methods for assessing DNA damage and DNA repair, strongly support the existence of measurable regions where genotoxic responses with increasing doses are insignificant relative to control. Model monofunctional alkylating agents have in vitro and in vivo datasets amenable to determination of points of departure (PoDs) for genotoxic effects. A session at the 2013 Society of Toxicology meeting provided an opportunity to survey the progress in understanding the biological basis of empirically-observed PoDs for DNA alkylating agents. Together with the literature published since, this review discusses cellular pathways activated by endogenous and exogenous alkylation DNA damage. Cells have evolved conserved processes that monitor and counteract a spontaneous steady-state level of DNA damage. The ubiquitous network of DNA repair pathways serves as the first line of defense for clearing of the DNA damage and preventing mutation. Other biological pathways discussed here that are activated by genotoxic stress include post-translational activation of cell cycle networks and transcriptional networks for apoptosis/cell death. The interactions of various DNA repair and DNA damage response pathways provide biological bases for the observed PoD behaviors seen with genotoxic compounds. Thus, after formation of DNA adducts, the activation of cellular pathways can lead to the avoidance of a mutagenic outcome. The understanding of the cellular mechanisms acting within the low-dose region will serve to better characterize risks from exposures to DNA-reactive agents at environmentally-relevant concentrations., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Adaptive Posttranslational Control in Cellular Stress Response Pathways and Its Relationship to Toxicity Testing and Safety Assessment.
- Author
-
Zhang Q, Bhattacharya S, Pi J, Clewell RA, Carmichael PL, and Andersen ME
- Subjects
- Animals, Humans, Oxidative Stress drug effects, Oxidative Stress physiology, Protein Processing, Post-Translational physiology, Signal Transduction drug effects, Signal Transduction physiology, Stress, Physiological physiology, Protein Processing, Post-Translational drug effects, Stress, Physiological drug effects, Toxicity Tests
- Abstract
Although transcriptional induction of stress genes constitutes a major cellular defense program against a variety of stressors, posttranslational control directly regulating the activities of preexisting stress proteins provides a faster-acting alternative response. We propose that posttranslational control is a general adaptive mechanism operating in many stress pathways. Here with the aid of computational models, we first show that posttranslational control fulfills two roles: (1) handling small, transient stresses quickly and (2) stabilizing the negative feedback transcriptional network. We then review the posttranslational control pathways for major stress responses-oxidative stress, metal stress, hyperosmotic stress, DNA damage, heat shock, and hypoxia. Posttranslational regulation of stress protein activities occurs by reversible covalent modifications, allosteric or non-allosteric enzymatic regulations, and physically induced protein structural changes. Acting in feedback or feedforward networks, posttranslational control may establish a threshold level of cellular stress. Sub-threshold stresses are handled adequately by posttranslational control without invoking gene transcription. With supra-threshold stress levels, cellular homeostasis cannot be maintained and transcriptional induction of stress genes and other gene programs, eg, those regulating cell metabolism, proliferation, and apoptosis, takes place. The loss of homeostasis with consequent changes in cellular function may lead to adverse cellular outcomes. Overall, posttranslational and transcriptional control pathways constitute a stratified cellular defense system, handling stresses coherently across time and intensity. As cell-based assays become a focus for chemical testing anchored on toxicity pathways, examination of proteomic and metabolomic changes as a result of posttranslational control occurring in the absence of transcriptomic alterations deserves more attention., (© The Author 2015. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
27. Profiling dose-dependent activation of p53-mediated signaling pathways by chemicals with distinct mechanisms of DNA damage.
- Author
-
Clewell RA, Sun B, Adeleye Y, Carmichael P, Efremenko A, McMullen PD, Pendse S, Trask OJ, White A, and Andersen ME
- Subjects
- Animal Use Alternatives, Apoptosis drug effects, Cell Culture Techniques, Cell Cycle drug effects, Cell Line, Cell Survival drug effects, Dose-Response Relationship, Drug, HCT116 Cells, High-Throughput Screening Assays, Humans, Mutagens chemistry, No-Observed-Adverse-Effect Level, Risk Assessment, Tumor Suppressor Protein p53 genetics, DNA Damage, Micronuclei, Chromosome-Defective chemically induced, Mutagens toxicity, Signal Transduction drug effects, Transcription, Genetic drug effects, Tumor Suppressor Protein p53 metabolism
- Abstract
As part of a larger effort to provide proof-of-concept in vitro-only risk assessments, we have developed a suite of high-throughput assays for key readouts in the p53 DNA damage response toxicity pathway: double-strand break DNA damage (p-H2AX), permanent chromosomal damage (micronuclei), p53 activation, p53 transcriptional activity, and cell fate (cell cycle arrest, apoptosis, micronuclei). Dose-response studies were performed with these protein and cell fate assays, together with whole genome transcriptomics, for three prototype chemicals: etoposide, quercetin, and methyl methanesulfonate. Data were collected in a human cell line expressing wild-type p53 (HT1080) and results were confirmed in a second p53 competent cell line (HCT 116). At chemical concentrations causing similar increases in p53 protein expression, p53-mediated protein expression and cellular processes showed substantial chemical-specific differences. These chemical-specific differences in the p53 transcriptional response appear to be determined by augmentation of the p53 response by co-regulators. More importantly, dose-response data for each of the chemicals indicate that the p53 transcriptional response does not prevent micronuclei induction at low concentrations. In fact, the no observed effect levels and benchmark doses for micronuclei induction were less than or equal to those for p53-mediated gene transcription regardless of the test chemical, indicating that p53's post-translational responses may be more important than transcriptional activation in the response to low dose DNA damage. This effort demonstrates the process of defining key assays required for a pathway-based, in vitro-only risk assessment, using the p53-mediated DNA damage response pathway as a prototype., (© The Author 2014. Published by Oxford University Press on behalf of Toxicological Sciences.)
- Published
- 2014
- Full Text
- View/download PDF
28. Use of mode of action data to inform a dose-response assessment for bladder cancer following exposure to inorganic arsenic.
- Author
-
Gentry PR, Yager JW, Clewell RA, and Clewell HJ 3rd
- Subjects
- Animals, Arsenic pharmacokinetics, Arsenic standards, Carcinogens pharmacokinetics, Carcinogens standards, Dose-Response Relationship, Drug, Gene Expression Regulation, Humans, Mice, Risk Assessment, Urinary Bladder Neoplasms epidemiology, Urinary Bladder Neoplasms metabolism, Water Pollutants, Chemical pharmacokinetics, Water Pollutants, Chemical standards, Arsenic toxicity, Carcinogens toxicity, Urinary Bladder Neoplasms chemically induced, Water Pollutants, Chemical toxicity
- Abstract
In the recent National Research Council report on conducting a dose-response assessment for inorganic arsenic, the committee remarked that mode of action data should be used, to the extent possible, to extrapolate below the observed range for epidemiological studies to inform the shape of the dose-response curve. Recent in vitro mode of action studies focused on understanding the development of bladder cancer following exposure to inorganic arsenic provide data to inform the dose-response curve. These in vitro data, combined with results of bladder cancer epidemiology studies, inform the dose-response curve in the low-dose region, and include values for both pharmacokinetic and pharmacodynamic variability. Integration of these data provides evidence of a range of concentrations of arsenic for which no effect on the bladder would be expected. Specifically, integration of these results suggest that arsenic exposures in the range of 7-43 ppb in drinking water are exceedingly unlikely to elicit changes leading to key events in the development of cancer or noncancer effects in bladder tissue. These findings are consistent with the lack of evidence for bladder cancer following chronic ingestion of arsenic water concentrations <100 ppb in epidemiological studies., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
29. A map of the PPARα transcription regulatory network for primary human hepatocytes.
- Author
-
McMullen PD, Bhattacharya S, Woods CG, Sun B, Yarborough K, Ross SM, Miller ME, McBride MT, LeCluyse EL, Clewell RA, and Andersen ME
- Subjects
- Binding Sites, Chromosome Mapping, Down-Regulation, Humans, Microarray Analysis, PPAR alpha chemistry, PPAR alpha metabolism, Signal Transduction, Transcription, Genetic, Up-Regulation, Computational Biology, Dose-Response Relationship, Drug, Hepatocytes physiology, PPAR alpha genetics
- Abstract
Nuclear receptor activation in liver leads to coordinated alteration of the expression of multiple gene products with attendant phenotypic changes of hepatocytes. Peroxisome proliferators including endogenous fatty acids, environmental chemicals, and drugs induce a multi-enzyme metabolic response that affects lipid and fatty acid processing. We studied the signaling network for the peroxisome proliferator-associated receptor alpha (PPARα) in primary human hepatocytes using the selective PPARα ligand, GW7647. We measured gene expression over multiple concentrations and times and conducted ChIP-seq studies at 2 and 24h to assess genomic binding of PPARα. Over all treatments there were 192 genes differentially expressed. Of these only 51% showed evidence of PPARα binding-either directly at PPARα response elements or via alternative mechanisms. Almost half of regulated genes had no PPARα binding. We then developed two novel bioinformatics methods to visualize the dose-dependent activation of both the transcription factor circuitry for PPARα and the downstream metabolic network in relation to functional annotation categories. Available databases identified several key transcription factors involved with the non-genomic targets after GW7647 treatment, including SP1, STAT1, ETS1, ERα, and HNF4α. The linkage from PPARα binding through gene expression likely requires intermediate protein kinases to activate these transcription factors. We found enrichment of functional annotation categories for organic acid metabolism and cell lipid metabolism among the differentially expressed genes. Lipid transport processes showed enrichment at the highest concentration of GW7647 (10 μM). While our strategy for mapping transcriptional networks is evolving, these approaches are necessary in moving from toxicogenomic methods that derive signatures of activity to methods that establish pathway structure, showing the coordination of the activated nuclear receptor with other signaling pathways., (Copyright © 2013 The Authors. Published by Elsevier Ireland Ltd.. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
30. Dose-response modeling of etoposide-induced DNA damage response.
- Author
-
Li Z, Sun B, Clewell RA, Adeleye Y, Andersen ME, and Zhang Q
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Survival drug effects, Computer Simulation, Flow Cytometry, Humans, Stochastic Processes, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents, Phytogenic toxicity, DNA Breaks, Double-Stranded drug effects, Dose-Response Relationship, Drug, Etoposide toxicity, Models, Biological
- Abstract
The 2007 National Research Council Report "Toxicity Testing in the 21st Century: A Vision and A Strategy" recommended an integrated, toxicity pathway-oriented approach for chemical testing. As an integral component of the recommendation, computational dose-response modeling of toxicity pathways promises to provide mechanistic interpretation and prediction of adverse cellular outcomes. Among the many toxicity pathways, the DNA damage response is better characterized and thus more suited for computational modeling. In the present study, we formulated a minimal mathematical model of this pathway to examine the dose response for etoposide (ETP), an anticancer drug that causes DNA double strand breaks (DSBs). In the model, DSB results from inhibition of topoisomerase by ETP and p53 is activated by a bistable switch composed of a positive feedback loop between ATM and γH2AX. Our stochastic model recapitulated the dose response for several molecular biomarkers measured with flow cytometry in HT1080 cells, including phosphorylated p53, ATM, γH2AX, and micronuclei. Model simulations were consistent with a bimodal pattern of p53 activation and a graded population-averaged response at high ETP concentrations. The graded response was a result of heterogeneous activation of individual cells due to molecular stochasticity. This work shows the value of combining data collection on single cell responses and mechanistic, stochastic modeling to develop and test hypothesis for the circuitry of important toxicity pathways. Future studies will determine how well this initial modeling effort agrees with a broader set of experimental studies on pathway responses by examining a more diverse group of DNA-damaging compounds.
- Published
- 2014
- Full Text
- View/download PDF
31. Evaluation of a predictive in vitro Leydig cell assay for anti-androgenicity of phthalate esters in the rat.
- Author
-
Balbuena P, Campbell J Jr, Clewell HJ 3rd, and Clewell RA
- Subjects
- Animals, Cell Line, Tumor, Leydig Cells metabolism, Male, Mice, Rats, Reproducibility of Results, Testosterone metabolism, Androgen Antagonists toxicity, Biological Assay methods, Leydig Cells drug effects, Phthalic Acids toxicity, Testosterone antagonists & inhibitors
- Abstract
An in vitro assay using the rat Leydig cell line R2C was evaluated for its ability to quantitatively predict inhibition of testosterone synthesis. Results obtained for endocrine active phthalates (MEHP, MBP), and inactive phthalates (MMP and MEP) were highly consistent with in vivo results based on tissue and media concentrations. Statistically significant inhibition of testosterone synthesis (p<0.05, 1-way ANOVA) was observed at 1 μM MBP and 3 μM MEHP, while MEP and MMP did not affect inhibition of testosterone synthesis until much higher concentrations (>> 100 μM). Concentrations causing 50% inhibition of testosterone synthesis for MBP and MEHP (3 and 6 μM respectively), were similar to in vivo values (3 and 7 μM). The R2C assay was used to determine the relative potency of 14 structurally diverse monoesters and oxidative metabolites of MEHP. Monoesters with alkyl chains 4-5 carbons in length had the highest potency for testosterone inhibition, while 0-2 carbon alkyl chains were least potent. Phase I metabolism did not completely inactivate MEHP, underscoring the need for metabolism data when interpreting in vitro pharmacodynamic data. This steroid inhibition assay provides a predictive in vitro alternative to expensive and timeconsuming developmental rat studies for phthalate-induced antiandrogenicity., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
32. Assessing dose-dependent differences in DNA-damage, p53 response and genotoxicity for quercetin and curcumin.
- Author
-
Sun B, Ross SM, Trask OJ, Carmichael PL, Dent M, White A, Andersen ME, and Clewell RA
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Dose-Response Relationship, Drug, Histones metabolism, Humans, Micronuclei, Chromosome-Defective chemically induced, Necrosis chemically induced, Oxidative Stress drug effects, Reactive Oxygen Species metabolism, Tumor Suppressor Protein p53 metabolism, Curcumin toxicity, DNA Damage, Mutagens toxicity, Oxidants toxicity, Quercetin toxicity
- Abstract
As part of a longer-term goal to create a quantitative mechanistic model of the p53-Mdm2 DNA-damage pathway, we are studying cellular responses to compounds causing DNA-damage by various modes-of action, including two natural polyphenols: quercetin (QUE) and curcumin (CUR). QUE and CUR are weak mutagens in some in vitro assays and possess both anti- or pro-oxidant effects depending on dose. This study examines the dose-response of DNA-damage pathway to these compounds in HT1080 cells (a human cell line with wild-type p53) at doses relevant to human exposure. CUR was more potent in causing reactive oxygen species, DNA damage (measured as phospho-H2AX) and p53 induction, with lowest observed effect levels (LOELs; 3-8 μM) approximately three-fold lower than QUE (20-30 μM). CUR showed a strong G2/M arrest and apoptosis at ≈ 10 μM. QUE caused S phase arrest at low doses (8 μM) and apoptosis was only induced at much higher doses (60 μM). At concentrations with similar levels of p-H2AX and p53 biomarkers, CUR caused greater micronuclei frequency. CUR induced clear increases micronuclei at 3-6 μM, while QUE had a weaker micronuclei response even at the highest doses. Thus, even with two compounds sharing common chemistries, DNA-damage response patterns differed significantly in terms of dose and cell fate., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
33. Disposition of diiosononyl phthalate and its effects on sexual development of the male fetus following repeated dosing in pregnant rats.
- Author
-
Clewell RA, Sochaski M, Edwards K, Creasy DM, Willson G, and Andersen ME
- Subjects
- Amniotic Fluid metabolism, Animals, Endocrine Disruptors blood, Endocrine Disruptors pharmacokinetics, Female, Fetus drug effects, Fetus physiology, Liver metabolism, Male, No-Observed-Adverse-Effect Level, Phthalic Acids blood, Phthalic Acids pharmacokinetics, Placenta metabolism, Pregnancy, Rats, Rats, Sprague-Dawley, Testis metabolism, Testis pathology, Tissue Distribution, Endocrine Disruptors toxicity, Phthalic Acids toxicity, Sexual Development drug effects, Testis drug effects, Testosterone metabolism
- Abstract
Pregnant Sprague-Dawley rats received 50, 250, and 500 mg/kg/day diisononyl phthalate (DiNP) from GD 12 to 19 via corn oil gavage to study the dose response for effects on fetal male rat sexual development as well as metabolite disposition in the dam and fetus. Monoisononyl phthalate (MiNP), mono(carboxy-isooctyl) phthalate (MCiOP), mono(hydroxyl-isononyl) phthalate (MHiNP), mono(oxo-isononyl) phthalate (MOiNP), and monoisononyl phthalate glucuronide (MiNP-G) were found in all measured tissues. MCiOP was the major metabolite, followed in decreasing order by MiNP, MHiNP, MOiNP, and MiNP-G. Percentage of dose absorbed decreased at 750 mg/kg/day. Testosterone concentration in the fetal testes was reduced at 250 and 750 mg/kg/day. Multinucleated germ cells were increased in the testes of rats at 250 and 750 mg/kg/day. The no observed effect level (NOEL) for this study was 50 mg/kg/day based on increased MNGs and reduced testes testosterone concentration in the fetal rat., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
34. A dose response study to assess effects after dietary administration of diisononyl phthalate (DINP) in gestation and lactation on male rat sexual development.
- Author
-
Clewell RA, Thomas A, Willson G, Creasy DM, and Andersen ME
- Subjects
- Animals, Animals, Newborn, Dibutyl Phthalate blood, Dibutyl Phthalate pharmacokinetics, Dibutyl Phthalate toxicity, Diet, Dose-Response Relationship, Drug, Female, Lactation metabolism, Male, No-Observed-Adverse-Effect Level, Phthalic Acids blood, Phthalic Acids pharmacokinetics, Plasticizers pharmacokinetics, Pregnancy, Rats, Rats, Sprague-Dawley, Testis pathology, Phthalic Acids toxicity, Plasticizers toxicity, Sexual Development drug effects, Testis drug effects
- Abstract
Male rat sexual development was evaluated after dietary administration of 0, 760, 3800, 11,400 ppm diisononyl phthalate (DiNP) and 7600 ppm dibutyl phthalate (DBP) from gestation day (GD) 12 to postnatal day (PND) 14. Maternal weight was reduced on GD 20, PND 2 and 14 at 11,400 ppm DiNP. Pup weight was reduced on PND 2 and 14 at 11,400 and 3800 ppm DiNP. DBP induced multinucleated germ cells (MNGs) and Leydig cell aggregates (LCAs) in PND 2 testes. 7600 ppm DBP reduced anogenital distance (AGD) on PND 2 and 14, and increased nipple retention and reproductive tract malformations on PND 49. DiNP induced MNGs (3800 ppm) and LCAs (11,400 ppm) on PND 2, and reduced AGD (11,400 ppm) on PND 14. DiNP did not alter AGD, nipple retention or reproductive tract malformations on PND 49. Global endpoint analysis showed no evidence of a rat "phthalate syndrome" on PND 49 with DiNP administration., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
35. In vitro metabolism of di(2-ethylhexyl) phthalate (DEHP) by various tissues and cytochrome P450s of human and rat.
- Author
-
Choi K, Joo H, Campbell JL Jr, Clewell RA, Andersen ME, and Clewell HJ 3rd
- Subjects
- Animals, Biotransformation, Brain metabolism, Chromatography, Liquid, Diethylhexyl Phthalate analogs & derivatives, Female, Humans, Intestinal Mucosa metabolism, Kidney metabolism, Liver metabolism, Lung metabolism, Male, Rats, Rats, Inbred F344, Recombinant Proteins metabolism, Skin metabolism, Tandem Mass Spectrometry, Testis metabolism, Cytochrome P-450 Enzyme System metabolism, Diethylhexyl Phthalate metabolism, Plasticizers metabolism
- Abstract
In vitro metabolism of DEHP by subcellular fractions of human brain, intestine, kidney, liver, lung, skin, testis, rat liver and recombinant CYP isoforms of human and rat was investigated using LC-MS/MS. DEHP was rapidly hydrolyzed to mono(2-ethylhexyl) phthalate (MEHP) in 12 microsomal/cytosolic fractions of selected 7 human organs and rat liver but not in microsomal fractions of human brain and human female skin. MEHP was metabolized to CYP-mediated oxidative and dealkylated metabolites in human and rat liver and at a lower rate in human intestine. Measurable amounts of mono(2-ethyl-5-hydroxyhexyl) phthalate (5-OH MEHP), mono(2-ethyl-5-oxohexyl) phthalate (5-Oxo MEHP), mono(2-ethyl-5-carboxypentyl) phthalate (5-carboxy MEPP), mono(2-carboxymethyl-hexyl) phthalate (2-carboxy MMHP) and phthalic acid (PA) were formed by human liver fractions. Human CYP2C9(∗)1, CYP2C19 and rat CYP2C6 were the major CYP isoforms producing 5-OH MEHP and 5-Oxo MEHP metabolites; however, only human CYP2C9(∗)1 and 2C9(∗)2 produced 5-carboxy MEPP from MEHP. Additionally, human CYP3A4 and rat CYP3A2 were the primary enzymes for PA production via heteroatom dealkylation of MEHP. Percent total normalized rates (%TNR) by CYP2C9(∗)1 in human liver microsomes (HLM) were 94%, 98% and 100%, respectively, for 5-OH MEHP, 5-Oxo MEHP, 5-carboxy MEPP, and 76% for PA production by CYP3A4., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
36. Physiologically based pharmacokinetic/toxicokinetic modeling.
- Author
-
Campbell JL Jr, Clewell RA, Gentry PR, Andersen ME, and Clewell HJ 3rd
- Subjects
- Animals, Humans, Models, Theoretical, Styrene pharmacokinetics, Pharmacokinetics
- Abstract
Physiologically based pharmacokinetic (PBPK) models differ from conventional compartmental pharmacokinetic models in that they are based to a large extent on the actual physiology of the organism. The application of pharmacokinetics to toxicology or risk assessment requires that the toxic effects in a particular tissue are related in some way to the concentration time course of an active form of the substance in that tissue. The motivation for applying pharmacokinetics is the expectation that the observed effects of a chemical will be more simply and directly related to a measure of target tissue exposure than to a measure of administered dose. The goal of this work is to provide the reader with an understanding of PBPK modeling and its utility as well as the procedures used in the development and implementation of a model to chemical safety assessment using the styrene PBPK model as an example.
- Published
- 2012
- Full Text
- View/download PDF
37. Assessing the relevance of in vitro measures of phthalate inhibition of steroidogenesis for in vivo response.
- Author
-
Clewell RA, Campbell JL, Ross SM, Gaido KW, Clewell HJ 3rd, and Andersen ME
- Subjects
- Animals, Cell Survival drug effects, Down-Regulation drug effects, Female, Fetal Development drug effects, Fetus drug effects, Leydig Cells drug effects, Leydig Cells metabolism, Luteinizing Hormone pharmacology, Male, Mice, Pregnancy, Progesterone antagonists & inhibitors, Progesterone biosynthesis, Proteins metabolism, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Risk Assessment, Sexual Maturation drug effects, Structure-Activity Relationship, Testis drug effects, Testis embryology, Testosterone antagonists & inhibitors, Testosterone biosynthesis, Phthalic Acids toxicity, Steroids biosynthesis
- Abstract
Human phthalate exposure occurs as mixtures of diesters with varying activity towards testosterone-dependent development. Dibutyl (DBP), diethylhexyl (DEHP) and butylbenzyl (BBP) phthalate disrupt sexual development in the fetal rat. Dimethyl (DMP) and diethyl (DEP) phthalate do not. These differences in potency may result from differential delivery of the monophthalates to the testes or from variation in the abilities of the compounds to alter steroidogenesis. We tested five phthalates in pregnant rats (500mg/kg-d, GD12-19) and analyzed the fetal testes for corresponding monoesters (MMP, MEP, MBP, MEHP, MBeP). Testes MMP and MEP levels were 2-40-fold higher than the active monoesters, MBP and MEHP. BBP exposure led to low concentrations of MBeP, but similar MBP levels to DBP. An in vitro MA-10 cell assay measured the direct effect of monophthalates on testosterone production. MEHP inhibited LH-stimulated testosterone production at 1microM. RT-PCR confirmed down-regulation of genes associated with cholesterol transport and steroid synthesis and metabolism by MEHP. Five additional phthalates were tested for testosterone inhibition. MBP and mono-n-octyl phthalate were similar to MEHP; MMP, MEP and MBeP were poor inhibitors of testosterone production. Based on these results, differences in the phthalates' ability to interfere with sexual development in vivo appears to be more associated with differential potency for testosterone inhibition than differences in tissue doses.
- Published
- 2010
- Full Text
- View/download PDF
38. In utero exposure to chloroquine alters sexual development in the male fetal rat.
- Author
-
Clewell RA, Pluta L, Thomas RS, and Andersen ME
- Subjects
- Animals, Chloroquine toxicity, Female, Fetal Development drug effects, Fetal Development physiology, Male, Pregnancy, Prenatal Exposure Delayed Effects pathology, Rats, Rats, Sprague-Dawley, Sexual Development physiology, Testis drug effects, Testis embryology, Chloroquine analogs & derivatives, Prenatal Exposure Delayed Effects chemically induced, Sexual Development drug effects
- Abstract
Chloroquine (CQ), a drug that has been used extensively for the prevention and treatment of malaria, is currently considered safe for use during pregnancy. However, CQ has been shown to disrupt steroid homeostasis in adult rats and similar compounds, such as quinacrine, inhibit steroid production in the Leydig cell in vitro. To explore the effect of in utero CQ exposure on fetal male sexual development, pregnant Sprague-Dawley rats were given a daily dose of either water or chloroquine diphosphate from GD 16-18 by oral gavage. Chloroquine was administered as 200 mg/kg CQ base on GD 16, followed by two maintenance doses of 100 mg/kg CQ base on GD 16 and 18. Three days of CQ treatment resulted in reduced maternal and fetal weight on GD 19 and increased necrosis and steatosis in the maternal liver. Fetal livers also displayed mild lipid accumulation. Maternal serum progesterone was increased after CQ administration. Fetal testes testosterone, however, was significantly decreased. Examination of the fetal testes revealed significant alterations in vascularization and seminiferous tubule development after short-term CQ treatment. Anogenital distance was not altered. Microarray and RT-PCR showed down-regulation of several genes associated with cholesterol transport and steroid synthesis in the fetal testes. This study indicates that CQ inhibits testosterone synthesis and normal testis development in the rat fetus at human relevant doses.
- Published
- 2009
- Full Text
- View/download PDF
39. Kinetics of selected di-n-butyl phthalate metabolites and fetal testosterone following repeated and single administration in pregnant rats.
- Author
-
Clewell RA, Kremer JJ, Williams CC, Campbell JL, Sochaski MA, Andersen ME, and Borghoff SJ
- Subjects
- Amniotic Fluid metabolism, Animals, Area Under Curve, Biomarkers, Calibration, Chromatography, High Pressure Liquid, Dose-Response Relationship, Drug, Female, Gas Chromatography-Mass Spectrometry, Indicators and Reagents, Liver metabolism, Male, Mass Spectrometry, Phthalic Acids pharmacokinetics, Placenta metabolism, Pregnancy, Quality Control, Quinolines, Rats, Rats, Sprague-Dawley, Sexual Maturation drug effects, Testis drug effects, Testis embryology, Dibutyl Phthalate pharmacokinetics, Fetus metabolism, Testis metabolism, Testosterone metabolism
- Abstract
Human exposure to phthalic acid diesters occurs through a variety of pathways as a result of their widespread use in consumer products and plastics. Repeated doses of di-n-butyl phthalate (DBP) from gestation day (GD) 12 to 19 disrupt testosterone synthesis and male sexual development in the fetal rat. Currently little is known about the disposition of DBP metabolites, such as monobutyl phthalate (MBP) and its glucuronide conjugate (MBP-G), during gestation after repeated exposure to DBP. In order to gain a better understanding of the effect of repeated dosing on maternal and fetal metabolism and distribution, pregnant Sprague-Dawley rats were given a single dose of 500 mg/kg DBP on GD 19 or daily doses of 50, 100, and 500 mg/(kg day) from GD 12 to 19 via corn oil gavage. Dose-response evaluation revealed a non-linear increase in maternal and fetal plasma concentrations of MBP. Maternal and fetal MBP levels were slightly lower in animals after 8 days of dosing at 500 mg/(kg day). Fetal plasma MBP levels closely followed maternal plasma, while the appearance and elimination of MBP-G in fetal plasma were significantly delayed. MBP-G accumulated over time in the amniotic fluid. Inhibition of testosterone was rapid in fetal testes when exposed to DBP (500 mg/(kg day)) on GD 19. Within 24h, the level of inhibition in the fetus was similar between animals exposed to a single or multiple daily doses of 500 mg/(kg day). Examination of testosterone time-course data indicates a rapid recovery to normal levels within 24h post-dosing at DBP doses of 50 and 100 mg/(kg day), with a rebound to higher than normal concentrations at later time-points. MBP kinetics in fetal testes allows direct comparison of active metabolite concentrations and testosterone response in the fetal testes.
- Published
- 2009
- Full Text
- View/download PDF
40. Tissue exposures to free and glucuronidated monobutylyphthalate in the pregnant and fetal rat following exposure to di-n-butylphthalate: evaluation with a PBPK model.
- Author
-
Clewell RA, Kremer JJ, Williams CC, Campbell JL Jr, Andersen ME, and Borghoff SJ
- Subjects
- Administration, Oral, Animals, Area Under Curve, Female, Fetus drug effects, Male, Models, Biological, Pregnancy, Rats, Rats, Sprague-Dawley, Tissue Distribution, Dibutyl Phthalate pharmacokinetics, Fetus metabolism, Glucuronides metabolism, Maternal Exposure, Phthalic Acids metabolism
- Abstract
Human exposure to phthalic acid diesters occurs through a variety of pathways as a result of their widespread use in plastics. Repeated doses of di-n-butylphthalate (DBP) from gestation day (GD) 12 to 19 disrupt testosterone synthesis and male sexual development in the fetal rat. To gain a better understanding of the relationship of the target tissue (testes) dose to observed developmental effects, the pharmacokinetics of monobutyl phthalate (MBP) and its glucuronide (MBP-G) were examined in pregnant and fetal rats following single and repeated administration of DBP from GD 12-19. These data, together with results from previously published studies, were used to develop a physiologically based pharmacokinetic model for DBP and its metabolites in the male, pregnant and fetal rat. The model structure accounts for the major metabolic (hydrolysis, glucuronidation, oxidative metabolism) and transport processes (enterohepatic recirculation, urinary and fecal excretion, placental transfer). Extrapolation of the validated adult male rat model to gestation successfully predicts MBP and MBP-G levels in maternal plasma, placenta and urine, as well as the fetal plasma and testes. Sensitivity analysis indicates that plasma MBP kinetics are particularly sensitive to glucuronidation and enterohepatic recirculation: a decrease in the uridine 5'-diphospho-glucuronosyltransferase (UDPGT) capacity during gestation results in an increased MBP residence time, and saturation of UDPGT at the highest doses (> 100 mg/kg/day) causes a flattening out of the plasma time course data. Oxidative metabolism plays a significant role in elimination only at low doses (< 50 mg/kg DBP). Insights gained from modeling of the rat data will be used to support development of a human PBPK model for DBP.
- Published
- 2008
- Full Text
- View/download PDF
41. Development and specification of physiologically based pharmacokinetic models for use in risk assessment.
- Author
-
Clewell RA and Clewell HJ 3rd
- Subjects
- Animals, Humans, Reproducibility of Results, Risk Assessment, Uncertainty, Models, Biological, Pharmacokinetics
- Abstract
Risk assessments are performed to estimate the conditions under which individuals or populations may be harmed by exposure to environmental or occupational chemicals. In the absence of quantitative data in the human, this process is often dependent upon the use of animal and in vitro data to estimate human response. To reduce the uncertainty inherent in such extrapolations, there has been considerable interest in the development of physiologically based pharmacokinetic (PBPK) models of toxic chemicals for application in quantitative risk assessments. PBPK models are effective tools for integrating diverse dose-response and mechanistic data in order to more accurately predict human risk. Yet, for these models to be useful and trustworthy in performing the necessary extrapolations (species, doses, exposure scenarios), they must be thoughtfully constructed in accordance with known biology and pharmacokinetics, documented in a form that is transparent to risk assessors, and shown to be robust using diverse and appropriate data. This paper describes the process of PBPK model development and highlights issues related to the specification of model structure and parameters, model evaluation, and consideration of uncertainty. Examples are provided to illustrate approaches for selecting a "preferred" model from multiple alternatives.
- Published
- 2008
- Full Text
- View/download PDF
42. Perchlorate and radioiodide kinetics across life stages in the human: using PBPK models to predict dosimetry and thyroid inhibition and sensitive subpopulations based on developmental stage.
- Author
-
Clewell RA, Merrill EA, Gearhart JM, Robinson PJ, Sterner TR, Mattie DR, and Clewell HJ 3rd
- Subjects
- Adolescent, Child, Child, Preschool, Dose-Response Relationship, Drug, Drinking, Female, Fetus metabolism, Humans, Infant, Infant, Newborn, Lactation metabolism, Male, Mammary Glands, Human metabolism, Middle Aged, Milk, Human chemistry, Models, Biological, Placenta metabolism, Pregnancy, Iodine Radioisotopes pharmacokinetics, Maternal-Fetal Exchange, Perchlorates pharmacokinetics, Thyroid Gland metabolism, Water Pollutants, Chemical pharmacokinetics
- Abstract
Perchlorate (ClO4(-)) is a drinking-water contaminant, known to disrupt thyroid hormone homeostasis in rats. This effect has only been seen in humans at high doses, yet the potential for long term effects from developmental endocrine disruption emphasizes the need for improved understanding of perchlorate's effect during the perinatal period. Physiologically based pharmacokinetic/dynamic (PBPK/PD) models for ClO4(-) and its effect on thyroid iodide uptake were constructed for human gestation and lactation data. Chemical specific parameters were estimated from life-stage and species-specific relationships established in previously published models for various life-stages in the rat and nonpregnant adult human. With the appropriate physiological descriptions, these kinetic models successfully simulate radioiodide data culled from the literature for gestation and lactation, as well as ClO4(-) data from populations exposed to contaminated drinking water. These models provide a framework for extrapolating from chemical exposure in laboratory animals to human response, and support a more quantitative understanding of life-stage-specific susceptibility to ClO4(-). The pregnant and lactating woman, fetus, and nursing infant were predicted to have higher blood ClO4(-) concentrations and greater thyroid iodide uptake inhibition at a given drinking-water concentration than either the nonpregnant adult or the older child. The fetus is predicted to receive the greatest dose (per kilogram body weight) due to several factors, including placental sodium-iodide symporter (NIS) activity and reduced maternal urinary clearance of ClO4(-). The predicted extent of iodide inhibition in the most sensitive population (fetus) is not significant (approximately 1%) at the U.S. Environmental Protection Agency reference dose (0.0007 mg/kg-d).
- Published
- 2007
- Full Text
- View/download PDF
43. PBPK model for radioactive iodide and perchlorate kinetics and perchlorate-induced inhibition of iodide uptake in humans.
- Author
-
Merrill EA, Clewell RA, Robinson PJ, Jarabek AM, Gearhart JM, Sterner TR, and Fisher JW
- Subjects
- Female, Humans, Iodine Radioisotopes blood, Iodine Radioisotopes pharmacokinetics, Iodine Radioisotopes urine, Male, Perchlorates blood, Perchlorates urine, Predictive Value of Tests, Symporters metabolism, Thyroid Gland drug effects, Models, Biological, Perchlorates pharmacokinetics, Thyroid Gland metabolism, Water Supply standards
- Abstract
Detection of perchlorate (ClO4-) in several drinking water sources across the U.S. has lead to public concern over health effects from chronic low-level exposures. Perchlorate inhibits thyroid iodide (I-) uptake at the sodium (Na+)-iodide (I-) symporter (NIS), thereby disrupting the initial stage of thyroid hormone synthesis. A physiologically based pharmacokinetic (PBPK) model was developed to describe the kinetics and distribution of both radioactive I- and cold ClO4- in healthy adult humans and simulates the subsequent inhibition of thyroid uptake of radioactive I- by ClO4-. The model successfully predicts the measured levels of serum and urinary ClO4- from drinking water exposures, ranging from 0.007 to 12 mg ClO4-/kg/day, as well as the subsequent inhibition of thyroid 131I- uptake. Thyroid iodine, as well as total, free, and protein-bound radioactive I- in serum from various tracer studies, are also successfully simulated. This model's parameters, in conjunction with corresponding model parameters established for the male, gestational, and lactating rat, can be used to estimate parameters in a pregnant or lactating human, that have not been or cannot be easily measured to extrapolate dose metrics and correlate observed effects in perchlorate toxicity studies to other human life stages. For example, by applying the adult male rat:adult human ratios of model parameters to those parameters established for the gestational and lactating rat, we can derive a reasonable estimate of corresponding parameters for a gestating or lactating human female. Although thyroid hormones and their regulatory feedback are not incorporated in the model structure, the model's successful prediction of free and bound radioactive I- and perchlorate's interaction with free radioactive I- provide a basis for extending the structure to address the complex hypothalamic-pituitary-thyroid feedback system. In this paper, bound radioactive I- refers to I- incorporated into thyroid hormones or iodinated proteins, which may or may not be bound to plasma proteins.
- Published
- 2005
- Full Text
- View/download PDF
44. Evidence for competitive inhibition of iodide uptake by perchlorate and translocation of perchlorate into the thyroid.
- Author
-
Clewell RA, Merrill EA, Narayanan L, Gearhart JM, and Robinson PJ
- Subjects
- Animals, Binding, Competitive drug effects, Biotransformation, Humans, Perchlorates blood, Protein Binding, Symporters metabolism, Thyroid Gland chemistry, Thyroid Gland drug effects, Iodides metabolism, Perchlorates metabolism, Perchlorates pharmacology, Thyroid Gland metabolism
- Abstract
Various published data sets that investigate the potential effect of exogenous perchlorate (ClO4-) on the uptake of iodide in the thyroid and subsequent changes in thyroid hormone levels are available. In order to best use the data towards the prediction of human health effects resulting from ClO4- exposure, the available literature data must be integrated into a self-consistent, coherent, and parsimonious quantitative model based on the most likely mode of action of perchlorate effect on thyroid function. We submit that the simplest mode of action for ClO4- in the thyroid that remains consistent with all available data involves competitive inhibition of iodide transport into the thyroid follicle, transport of perchlorate into the thyroid follicle against a concentration gradient, further transport into the thyroid lumen (where it may again interfere with iodide transport), and, finally, passive diffusion back into the blood. We believe this description of perchlorate's kinetic behavior should serve as the foundation for predictive physiologically based pharmacokinetic (PBPK) models and as a working hypothesis for further experimental exploration.
- Published
- 2004
- Full Text
- View/download PDF
45. Predicting neonatal perchlorate dose and inhibition of iodide uptake in the rat during lactation using physiologically-based pharmacokinetic modeling.
- Author
-
Clewell RA, Merrill EA, Yu KO, Mahle DA, Sterner TR, Fisher JW, and Gearhart JM
- Subjects
- Animals, Animals, Suckling metabolism, Binding, Competitive drug effects, Female, Milk chemistry, Milk metabolism, Models, Biological, Perchlorates administration & dosage, Rats, Rats, Sprague-Dawley, Sodium Compounds administration & dosage, Symporters metabolism, Water Pollutants, Chemical administration & dosage, Animals, Newborn metabolism, Iodine Radioisotopes pharmacokinetics, Lactation metabolism, Perchlorates pharmacokinetics, Sodium Compounds pharmacokinetics, Water Pollutants, Chemical pharmacokinetics
- Abstract
Perchlorate (ClO4-), a contaminant in drinking water, competitively inhibits active uptake of iodide (I-) into various tissues, including mammary tissue. During postnatal development, inhibition of I- uptake in the mammary gland and neonatal thyroid and the active concentration ClO4- in milk indicate a potentially increased susceptibility of neonates to endocrine disruption. A physiologically based pharmacokinetic (PBPK) model was developed to reproduce measured ClO4- distribution in the lactating and neonatal rat and predict resulting effects on I- kinetics from competitive inhibition at the sodium iodide symporter (NIS). Kinetic I- and ClO4- behavior in tissues with NIS (thyroid, stomach, mammary gland, and skin) was simulated with multiple subcompartments, Michaelis-Menten (M-M) kinetics and competitive inhibition. Physiological and kinetic parameters were obtained from literature and experiment. Systemic clearance and M-M parameters were estimated by fitting simulations to tissue and serum data. The model successfully describes maternal and neonatal thyroid, stomach, skin, and plasma, as well as maternal mammary gland and milk data after ClO4- exposure (from 0.01 to 10 mg/kg-day ClO4-) and acute radioiodide (2.1 to 33,000 ng/kg I-) dosing. The model also predicts I- uptake inhibition in the maternal thyroid, mammary gland, and milk. Model simulations predict a significant transfer of ClO4- through milk after maternal exposure; approximately 50% to 6% of the daily maternal dose at doses ranging from 0.01 to 10.0 mg ClO4-/kg-day, respectively. Comparison of predicted dosimetrics across life-stages in the rat indicates that neonatal thyroid I- uptake inhibition is similar to the adult and approximately tenfold less than the fetus.
- Published
- 2003
- Full Text
- View/download PDF
46. Predicting fetal perchlorate dose and inhibition of iodide kinetics during gestation: a physiologically-based pharmacokinetic analysis of perchlorate and iodide kinetics in the rat.
- Author
-
Clewell RA, Merrill EA, Yu KO, Mahle DA, Sterner TR, Mattie DR, Robinson PJ, Fisher JW, and Gearhart JM
- Subjects
- Animals, Dose-Response Relationship, Drug, Drinking, Drug Interactions, Female, Iodides administration & dosage, Male, Models, Biological, Perchlorates administration & dosage, Pregnancy, Rats, Rats, Sprague-Dawley, Sodium Compounds administration & dosage, Thyroid Gland drug effects, Thyroid Gland metabolism, Water Supply, Fetus metabolism, Iodides pharmacokinetics, Maternal-Fetal Exchange, Perchlorates pharmacokinetics, Sodium Compounds pharmacokinetics
- Abstract
Perchlorate (ClO4-) disrupts endocrine homeostasis by competitively inhibiting the transport of iodide (I-) into the thyroid. The potential for health effects from human exposure to ClO4- in drinking water is not known, but experimental animal studies are suggestive of developmental effects from ClO4- induced iodide deficiency during gestation. Normal hormone-dependent development relies, in part, on synthesis of hormones in the fetal thyroid from maternally supplied iodide. Although ClO4- crosses the placenta, the extent of inhibition in the fetal thyroid is unknown. A physiologically-based pharmacokinetic (PBPK) model was developed to simulate ClO4- exposure and the resulting effect on iodide kinetics in rat gestation. Similar to concurrent model development for the adult male rat, this model includes compartments for thyroid, stomach, skin, kidney, liver, and plasma in both mother and fetus, with additional compartments for the maternal mammary gland, fat, and placenta. Tissues with active uptake are described with multiple compartments and Michaelis-Menten (M-M) kinetics. Physiological and kinetic parameters were obtained from literature and experiment. Systemic clearance, placental-fetal transport, and M-M uptake parameters were estimated by fitting model simulations to experimental data. The PBPK model is able to reproduce maternal and fetal iodide data over five orders of magnitude (0.36 to 33,000 ng/kg 131I-), ClO4- distribution over three orders of magnitude (0.01 to 10 mg/kg-day ClO4-) and inhibition of maternal thyroid and total fetal I- uptake. The model suggests a significant fetal ClO4- dose in late gestation (up to 82% of maternal dose). A comparison of model-predicted internal dosimetrics in the adult male, pregnant, and fetal rat indicates that the fetal thyroid is more sensitive to inhibition than that of the adult.
- Published
- 2003
- Full Text
- View/download PDF
47. PBPK predictions of perchlorate distribution and its effect on thyroid uptake of radioiodide in the male rat.
- Author
-
Merrill EA, Clewell RA, Gearhart JM, Robinson PJ, Sterner TR, Yu KO, Mattie DR, and Fisher JW
- Subjects
- Animals, Drug Therapy, Combination, Injections, Intravenous, Iodides administration & dosage, Iodine Radioisotopes, Male, Models, Biological, Perchlorates administration & dosage, Rats, Sodium Compounds administration & dosage, Symporters, Thyroid Gland drug effects, Iodides pharmacokinetics, Perchlorates pharmacokinetics, Sodium Compounds pharmacokinetics, Thyroid Gland metabolism
- Abstract
Due to perchlorate's (ClO4-) ability to competitively inhibit thyroid iodide (I-) uptake through the sodium-iodide symporter (NIS), potential human health risks exist from chronic exposure via drinking water. Such risks may include hypothyroidism, goiter, and mental retardation (if exposure occurs during critical periods in neurodevelopment). To aid in predicting perchlorate's effect on normal I- kinetics, we developed a physiologically-based pharmacokinetic (PBPK) model for the adult male rat. The model structure describes simultaneous kinetics for both anions together with their interaction at the NIS, in particular, the inhibition of I- uptake by ClO4-. Subcompartments and Michaelis-Menten (M-M) kinetics were used to describe active uptake of both anions in the thyroid, stomach, and skin. Separate compartments for kidney, liver, plasma, and fat were described by passive diffusion. The model successfully predicts both 36ClO4- and 125I- kinetics after iv doses of 3.3 mg/kg and 33 mg/kg, respectively, as well as inhibition of thyroid 125I- uptake by ClO4- after iv doses of ClO4- (0.01 to 3.0 mg/kg). The model also predicts serum and thyroid ClO4- concentrations from 14-day drinking water exposures (0.01 to 30.0 mg ClO4-/kg/day) and compensation of perchlorate-induced inhibition of radioiodide uptake due to upregulation of the thyroid. The model can be used to extrapolate dose metrics and correlate observed effects in perchlorate toxicity studies to other species and life stages, such as rat gestation (Clewell et al., 2003). Because the model successfully predicts perchlorate's interaction with iodide, it provides a sound basis for future incorporation of the complex hypothalamic-pituitary-thyroid feedback system.
- Published
- 2003
- Full Text
- View/download PDF
48. Pharmacokinetics of toxic chemicals in breast milk: use of PBPK models to predict infant exposure.
- Author
-
Clewell RA and Gearhart JM
- Subjects
- Adult, Breast Feeding, Chemical Phenomena, Chemistry, Physical, Female, Forecasting, Humans, Infant, Infant, Newborn, Iodides pharmacokinetics, Kinetics, Male, Perchlorates pharmacokinetics, Risk Assessment, Sodium Compounds pharmacokinetics, Environmental Pollutants pharmacokinetics, Environmental Pollutants toxicity, Milk, Human chemistry, Models, Chemical
- Abstract
Factors controlling the transfer of potentially toxic chemicals in the breast milk of nursing mothers include both chemical characteristics, such as lipophilicity, and physiologic changes during lactation. Physiologically based pharmacokinetic (PBPK) models can aid in the prediction of infant exposure via breast milk. Benefits of these quantitative models include the ability to account for changing maternal physiology and transfer kinetics, as well as the chemical-specific characteristics, in order to produce more accurate estimates of neonatal risk. A recently developed PBPK model for perchlorate and iodide kinetics in the lactating and neonatal rat demonstrates the utility of PBPK modeling in predicting maternal and neonatal distribution of these two compounds. This model incorporates time-dependent changes in physiologic characteristics and includes interactions between iodide and perchlorate that alter the distribution and kinetics of iodide.
- Published
- 2002
- Full Text
- View/download PDF
49. The use of physiologically based models to integrate diverse data sets and reduce uncertainty in the prediction of perchlorate and iodide kinetics across life stages and species.
- Author
-
Clewell RA, Merrill EA, and Robinson PJ
- Subjects
- Age Factors, Animals, Female, Forecasting, Humans, Iodides pharmacokinetics, Kinetics, Lactation, Male, Perchlorates pharmacokinetics, Rats, Reproducibility of Results, Risk Assessment, Sodium Compounds pharmacokinetics, Models, Biological, Perchlorates adverse effects, Sodium Compounds adverse effects, Water Supply
- Abstract
The effects of perchlorate on the incorporation of iodide into thyroid hormones have been studied for more than 40 years in many species and under varying exposure conditions. Nevertheless, the database for this drinking water contaminant is still incomplete, particularly with regard to human developmental risk. A method for integrating the available data and forming meaningful conclusions for risk assessment is needed. To this end, an initial suite of physiologically based pharmacokinetic (PBPK) models has been developed, which incorporates physiological data for the relevant species and life stages and kinetic data for perchlorate and iodide, as well as the interaction between the two anions. The validated models successfully describe perchlorate-induced inhibition of thyroid iodide uptake and perchlorate and iodide kinetics in the male, pregnant, lactating, fetal, and neonatal rats and the adult humans. The relationships of model-predicted internal dose metrics and kinetic parameters allow a direct comparison of internal dose metrics across life stages in rats and humans. By incorporating all the available data, these models provide a framework for species and life stage extrapolation where the lack of specific data sets would otherwise limit predictive capability. This paper demonstrates two approaches for calculating life stage-specific equivalent doses in a risk assessment for perchlorate: the direct combination of validated model predictions, and the development of preliminary PBPK models for the human-sensitive populations based on the relationship of the parameters in the validated rat and human models. Either approach can be used to perform the needed dosimetry. However, the second approach provides the advantage of a preliminary human life stage-specific PBPK model that can be used for identification of key data gaps and estimation of uncertainty.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.